• Non ci sono risultati.

MicroRNAs in liver cancer: a model for investigating pathogenesis and novel therapeutic approaches

N/A
N/A
Protected

Academic year: 2021

Condividi "MicroRNAs in liver cancer: a model for investigating pathogenesis and novel therapeutic approaches"

Copied!
12
0
0

Testo completo

(1)

Review

MicroRNAs in liver cancer: a model for investigating

pathogenesis and novel therapeutic approaches

E Callegari

1

, L Gramantieri

2

, M Domenicali

3

, L D’Abundo

1

, S Sabbioni*

,4

and M Negrini*

,1

MicroRNAs (miRNAs) constitute a large class of short RNAs (e.g., 20–24 nucleotides in length), whose main function is to

posttranscriptionally regulate the expression of protein-coding genes. Their importance in tumorigenesis has been

demonstrated over the past decade, and correspondingly, they have emerged as potential therapeutic molecules and targets.

Liver cancer is one of the most common neoplastic diseases worldwide, and it currently has a poor prognosis owing to largely

ineffective therapeutic options. Liver cancer is also an excellent model for testing miRNA-based therapy approaches as it can be

easily targeted with the systemic delivery of oligonucleotides. In recent years, the role of miRNAs in hepatocellular carcinoma

(HCC) has been established with molecular studies and the development of animal models. These studies have also provided the

basis for evaluating the therapeutic potential of miRNAs, or anti-miRNAs. In general, the safety of miRNAs has been proven and

antitumor activity has been observed. Moreover, because of the absence or presence of mild side effects, the prophylactic use of

miRNA-based approaches may be foreseen.

Cell Death and Differentiation (2015) 22, 46–57; doi:10.1038/cdd.2014.136; published online 5 September 2014

Facts

 Hepatocellular carcinoma (HCC) is a primary malignant

disease of the liver with poor prognosis.

 With the exception of the multi-kinase inhibitor, sorafenib,

there are no effective systemic treatments available for

HCC.

 Several miRNAs are differentially expressed in HCC and

may be pathogenically relevant.

 Genetically modified animal models have been developed

to demonstrate the direct role of miRNAs in the initiation

and/or progression of cancer.

 The in vivo therapeutic efficacy of certain miRNA mimics or

anti-miRNA oligonucleotides has been evaluated.

Open Questions

 Do the animal models that are available faithfully represent

the etiology and natural history of human HCC?

 Are methods for the in vivo systemic delivery of miRNAs/

anti-miRNAs sufficiently developed, and can they be

optimized?

 Are miRNA/anti-miRNA molecules effective against liver

cancer?

 What are the clinical settings in which miRNA/anti-miRNA

molecules may be successfully applied?

MicroRNAs (miRNAs) constitute a large class of short RNAs

(e.g., 20–24 nucleotides in length), which have key roles in cell

development and differentiation by mediating the

post-transcriptional regulation of protein-coding genes.

1

Over the

past decade, the critical role of miRNAs in tumorigenesis has

been widely investigated and their important regulatory action

in several biological processes that are frequently altered in

cancer has been described. At present, miRNAs have a

well-recognized role in human carcinogenesis, including

hepato-carcinogenesis, and accumulating experimental evidence

indicates that they may act as oncogenes or tumor suppressor

genes.

2,3

It is also well known that the expression of several

miRNAs is deregulated in human HCC compared with normal

tissue.

4,5

In this context, miRNAs have also emerged as novel

molecules or targets for tumor therapy, and liver cancer

represents an excellent model for their testing.

HCC is the most common primary liver malignant disease,

and the third cause of cancer-related deaths, worldwide.

6

It has a poor prognosis and the therapeutic options currently

available are largely ineffective. Molecular studies have

revealed several pathogenic mechanisms at the basis of liver

cancer. For example, HCC pathogenesis involves multiple

1

Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, Universita` di Ferrara, 44121 Ferrara, Italy;

2

Centro di Ricerca Biomedica Applicata e Dipartimento di

Medicina Interna, Policlinico S. Orsola-Malpighi e Universita` di Bologna, 40138 Bologna, Italy;

3

Unita` di Semeiotica Medica, Policlinico S. Orsola-Malpighi, Dipartimento

di Scienze Mediche e Chirurgiche, Universita` di Bologna, 40138 Bologna, Italy and

4

Dipartimento di Scienze della Vita e Biotecnologie, Universita` di Ferrara, 44121

Ferrara, Italy

*Corresponding authors: S Sabbioni, Dipartimento di Scienze della Vita e Biotecnologie, Universita` di Ferrara, 44121 Ferrara, Italy. Tel: +39 0532 455319;

Fax: +39 0532 455875; E-mail: sbs@unife.it

or M Negrini, Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, Universita` di Ferrara, 44121 Ferrara, Italy. Tel: +39 0532 455399; Fax: +39 0532 455875;

E-mail: ngm@unife.it

Received 17.3.14; revised 02.7.14; accepted 24.7.14; Edited by RA Knight; published online 05.9.14

Abbreviations: HCC, hepatocellular carcinoma; HCV, hepatitis C virus; miRNA, microRNA; DEN, N-nitrosodiethylamine; HGF, hepatocyte growth factor; CDK,

cyclin-dependent kinase; IAP, inhibitor of apoptosis protein; ROCK2, rho-cyclin-dependent kinase; AAV, adeno-associated virus; CRAds, conditionally replicating adenovirus

(2)

genetic and epigenetic alterations that lead to the deregulation

of several signaling pathways, including the p53, PI3K/Akt/

mTOR, Ras/Raf/MEK/ERK, IGFR, and Wnt/b-catenin

path-ways (for reviews, see Gramantieri et al.

7

and Aravalli et al.

8

).

Recent studies using next-generation sequencing technology

have further refined our knowledge of the signaling pathways

involved in liver carcinogenesis.

9–11

Regulation of these

cellular signaling pathways by miRNAs indicates that this

class of short RNAs may present a new set of potentially

therapeutic tools for HCC.

12,13

miRNAs and HCC

Several studies have revealed that the expression of certain

miRNAs is deregulated in human HCC compared with

matched nonneoplastic tissue (for a comprehensive review,

see Negrini et al.

14

). Moreover, molecular and cellular studies

have established that aberrantly expressed miRNAs can

affect crucial cancer-associated pathways.

2,3

Cell proliferation and survival. The discovery of

modu-lated gene targets has helped recognize the contribution

of miRNAs to cancer-associated pathways. Unrestricted

cellular proliferation and prolonged survival have critical

roles in the process of hepatocarcinogenesis. In the cell cycle

context, abnormal expression of miRNAs has been shown to

alter the functionality of several cell cycle regulators,

including RB1, cyclin-dependent kinases (CDKs), cyclins,

and CDK inhibitors. RB1 is a target of miR-335.

15

miR-335 is

downregulated during mesenchymal stem cell

differentia-tion

16

and upregulated by the Wnt signaling pathway,

16

which is often activated in HCC. RB1 is also a target of

miR-221, a miRNA that is frequently overexpressed in HCC.

17,18

These observations suggest that expression of RB1, a key

G1–S cell cycle transition checkpoint protein, may be

aberrantly regulated by altered miRNA expression. Among

the other proteins that control cell cycle progression

(Figure 1), cyclins are reported to be upregulated in HCC,

whereas negative cell cycle regulators are often

down-regulated compared with surrounding parenchyma.

19

Over-expression of cyclinD1/CDK4 has been detected in

B60% of

HCC cases.

20

Among CDK inhibitors, p16/INK4A is

function-ally inactivated owing to deletions in the short arm of

chromosome 9 in about 20% of HCCs

21

or by promoter

methylation in 30–70% of HCC cases.

22

All of these genes

are also reported to be affected by altered expression of

miRNAs (Figure 1). For example, miR-124 and miR-203,

which inhibit the growth of HCC cells via downregulation of

CDK6,

23

are methylated and silenced in HCC cell lines and

primary tumors.

23

Direct targeting of cyclin D1, CDK6, and

E2F3 by miR-195 can also block the G1–S transition,

whereas inhibition of miR-195 promotes cell-cycle

progres-sion.

24

MiR-195 can also suppress the ability of HCC cells to

form colonies in vitro, and the development of tumors in nude

mice. In most HCC tissues and cell lines, miR-195

expres-sion is reduced,

24

and this may contribute to the upregulation

of cyclinD1/CDK that is observed in HCC. MiR-26 is another

miRNA whose expression is reduced in HCC,

25

thereby

preventing its induction of cell cycle arrest in liver cancer cells

via the direct targeting of cyclins D2 and E2.

26

Members of

the KIP family of CDK inhibitors, CDKN1A/p21, CDKN1B/

p27, and CDKN1C/p57, also act as tumor suppressors in

HCC

by

negatively

affecting

cell-cycle

progression.

CDKN1B/p27 and CDKN1C/p57 proteins are downregulated

in HCC compared with surrounding cirrhosis.

27

One

mechan-ism that leads to a reduction in CDKN1C/p57 expression is

the loss of maternal allele methylation at the

KvDMR1-imprinted locus at 11p15.5. This has been found in 20–50%

of HCCs.

28

Another mechanism involved in the

downregula-tion of p27 and p57 is the overexpression of miR-221/222,

which occurs in about 70–80% of HCCs.

18

Owing to their

inhibition of RB1 expression as well, levels of miR-221/222

appear to be major factors in cell-cycle control. CDKN1A/p21

is downregulated by oncogenic members of the miR-17-92

family.

29–33

Among the three clusters of the miR-17-92 family

of the human genome, those on chromosomes 13 and 7 are

upregulated in HCC.

34

Disruption of apoptosis in HCC has been extensively

reviewed.

35,36

In this context, miRNAs contribute to the

altered expression of members of the BCL2 family

(Figure 2). For example, upregulation of antiapoptotic

members may result from reduced expression of miRNAs,

with miR-122 controlling BCL-W and BCL-XL

37

while the latter

is also controlled by let-7 members.

38

MCL1 is a target of

miR-101,

39

miR-193b,

40

and miR-29,

41,42

and all of these miRNAs

are downregulated in HCC. Conversely, proapoptotic

mem-bers may be inhibited by overexpressed miRNAs. For

example, the BH3-only proteins, BMF and BID, are targeted

by miR-221 and miR-25, respectively.

29,30,43

These

mechan-isms have the potential to allow miR-221 to protect cells from

‘anoikis’, a form of apoptosis induced by the detachment of

anchorage-dependent cells from the surrounding extracellular

matrix. MiR-25 may also impair the TGF-b tumor suppressor

pathway. Both pathways represent critical steps in the

process of metastasis.

44,45

miR-483 is a miRNA located

within intron 2 of the IGF2 gene, and it is highly expressed in

fetal liver, yet is barely detectable in adult liver.

46

Moreover, it

is overexpressed in 30–40% of HCCs either independently or

in combination with IGF2. MiR-483-3p has also been shown to

promote cell survival by repressing translation of the

p53-inducible BH3-only protein, PUMA/BBC3.

47

Apoptosis is also controlled by molecules not belonging to

the BCL2 family. For example, the inhibitor of apoptosis

protein (IAP), survivin, is overexpressed in HCC and is

controlled by miR-203,

48

a miRNA silenced by aberrant DNA

methylation in HCC tissue.

23

Another miRNA, miR-21,

appears to promote cell survival under stressful conditions.

Indeed, miR-21 is upregulated in HCC, as well as in many

other human neoplasms, and is associated with poor

prognosis

49,50

and resistance to chemotherapy.

51–53

These

effects appears to be linked to the survival advantage

imparted by miR-21 via the direct targeting of proapoptotic

genes, such as the tumor suppressor lipid-phosphatase,

PTEN, which controls the phosphatidyl inositol 3-phosphate

kinase (PI3K) signaling pathway

54–56

and programmed cell

death 4 (Pdcd4) protein.

53,57–62

The latter also has a role in

TGF-b-induced apoptosis.

At the nexus of cell cycle and apoptosis signaling is a

molecular pathway that is controlled by the tumor-suppressor

protein, p53. Genetic alterations in the TP53 gene have been

(3)

Figure 1

Aberrant miRNA expression in liver cancer promotes cell-cycle progression. A simplified pathway of cell cycle regulation is presented that shows the effects of various aberrantly expressed miRNAs in liver cancer. Upregulated miRNAs are indicated with a red up arrow, and downregulated miRNAs are indicated with a green down arrow. Upregulated miRNAs exert a stronger inhibitory effect on their targets, whereas downregulated miRNAs mediate weaker effects on their targets. Taken together, multiple aberrant miRNAs act on this pathway to support cell cycle progression

Figure 2

Aberrant miRNA expression in liver cancer affects the functionality of apoptosis pathways. A simplified apoptosis pathway is presented that shows the effects of various miRNAs that are aberrantly expressed in liver cancer. The aberrant expression of multiple miRNAs supports cell survival

(4)

extensively described in HCC,

21,63,64

and execution of the p53

pathway in HCC is affected by miRNA expression at various

levels (Figure 2). For example, the loss of miR-122 expression

in HCC cells leads to repression of p53 via upregulation of

cyclin G1.

65,66

MiR-145 also induces p53 activity by an

unknown mechanism,

67

and is commonly downregulated in

HCC.

65

In many cases, miRNAs that are downstream

effectors of p53 are downregulated owing to p53 loss of

function. Although not fully proven in HCC, this situation is

exemplified by miR-34a that is downregulated in some HCCs,

most likely owing to the presence of a nonfunctional p53.

However, in other HCCs, miR-34a is upregulated.

54,68

Lack of

miR-34a induction can promote several biological effects

through increased expression of CDK4, cyclin E2, BCL2, and

the tyrosine kinase receptor, MET.

69–74

Other examples of

p53-inducible miRNAs include miR-145, whose lack of

expression leads to the activation of MYC,

75

or the cluster,

miR-192/215.

76,77

Similar to 34, either 145 or

miR-192/215 can promote cell-cycle arrest and/or apoptosis,

colony

suppression,

and

can

increase

CDKN1A/p21

levels.

67,76,77

Invasion and metastasis. Advanced tumor features, such

as the ability of cancer cells to promote uncontrolled

angiogenesis and to invade tissues and blood vessels, are

also affected by miRNA deregulation. One of the signaling

pathways that confers invasive potential to HCC cells is

mediated by the HGF/MET axis. Hepatocyte growth factor

(HGF) binds to its transmembrane tyrosine kinase receptor,

MET, and promotes hepatocyte proliferation, migration,

survival, and angiogenesis. HGF also mediates invasive

growth

during

embryonic

development

and

tumori-genesis.

78,79

Overexpression of MET is found in 40–70% of

HCCs

80–83

and is regulated by miR-199-3p (indicated earlier

as miR-199a*), miR-34a, miR-23b, and miR-1.

84–88

All these

miRNAs are downregulated in HCC, thereby contributing to

the upregulation of c-MET. Among the downstream effectors

of RTKs, the overexpression of RAS, but rarely activating

point mutations, has been demonstrated in HCC.

89,90

All

members of the RAS family have been shown to be

modulated by various members of the let-7 family, and the

latter are downregulated in HCC,

65

as well as in several other

human cancers,

91–97

thus suggesting that let-7 potentially

contributes to the upregulation of RAS.

As previously mentioned, the PI3K/AKT/PTEN pathway

promotes cell survival as well as invasion and metastasis.

Correspondingly, the targeting of PTEN by 21 and

miR-221 can favor an invasive phenotype.

98,99

Indeed, the

silencing of PTEN and PDCD4 by miR-21 has resulted in a

decrease in apoptosis and an increase in cell invasion.

MiR-21, which is upregulated in HCC and in most human

malignancies, is linked to invasion and metastasis via its

targeting of multiple tumor/metastasis suppressor genes,

including tropomyosin 1 (TPM1), maspin, tissue inhibitor of

metalloproteinase 3 (TIMP3) gene, and RHOB.

54,100–103

In

addition, miR-21-mediated inhibition of RECK, a

membrane-anchored glycoprotein that negatively regulates matrix

metal-loproteinase-9, leads to increased cell invasion.

98

In the

context of the PI3K/AKT pathway, activation of the mTOR

pathway has been found to be common to several human

cancers, including HCC.

104

Moreover, when it is

over-expressed, it is associated with poor prognosis, invasion,

and metastasis.

105

Despite this, a very low rate of genetic

alterations that affect the mTOR pathway in HCC has been

reported.

104

Recently, mTOR was identified as a direct target

of 3p, and to be inversely correlated with

miR-199a-3p in HCC.

84

The miR-199/214 cluster is of particular interest

as it is downregulated in the majority of HCCs that have been

studied,

4,54,65,84,106,107

as well as in other human

malignan-cies,

108,109

in cancer-derived cell lines,

110

and in experimental

neoplastic and pre-neoplastic conditions.

111

Other deregulated miRNAs that have a role in the invasive

and metastatic properties of HCC cells include 122,

miR-139, and miR-151. For example, loss of miR-122 enhances

cell migration and invasion, and its restoration in metastatic

liver cancer cells has been shown to significantly reduce

migration, invasion, and anchorage-independent growth

in vitro, as well as tumorigenesis, angiogenesis, and

intrahepatic metastasis in vivo.

66,112,113

Invasive and

meta-static properties of HCC are also linked to the loss of control of

ADAM17 by miR-122. Indeed, specific silencing of ADAM17

resulted in a dramatic reduction in migration and invasion

in vitro, and reduced tumorigenesis, angiogenesis, and local

invasion in the livers of nude mice.

114

These observations are

similar to those associated with the restoration of miR-122.

Downregulation of miR-122, which has been detected in about

70% of HCCs,

5,54,65

is also associated with the development

of intrahepatic metastases

114

and a shorter time to

recur-rence.

66

Reduced expression of miR-139 has also been

detected in HCC,

106,115

and has specifically been linked to the

acquisition of metastatic properties.

115

In HCC cells,

over-expression of miR-139 has significantly reduced cell migration

and invasion in vitro, and the incidence and severity of lung

metastases in vivo. These effects were linked to interactions

between miR-139 and the 3

0

untranslated region of rho-kinase

2 (ROCK2). In human metastatic HCC, expression of miR-139

is reduced and levels of miR-139 inversely correlate with

levels of ROCK2 protein in human HCC samples.

115

MiR-151

is frequently amplified and overexpressed in HCC in

combination with its host gene, focal adhesion kinase

(FAK).

116

It is possible that the host gene, FAK, cooperates

with miR-151 to enhance cell motility and spreading effects,

and this would suggest that miR-151 has a critical role in tumor

invasion and metastasis. MiR-151-5p targeting of RhoGDIA, a

putative metastasis suppressor, has also been found to

increase HCC cell migration and invasion, as well as the

activation of Rac1, Cdc42, and Rho GTPases.

116

The b-catenin pathway has a central role in the

develop-ment of liver cancer and is an essential component of both

intercellular junctions and canonical Wnt signaling. Thus,

b-catenin is important for the regulation of cell proliferation,

differentiation, and stemness

19,117–121

(Figure 3). In HCC, the

Wnt/b-catenin pathway has been found to be abnormally

activated through gain-of-function mutations at the

N-termi-nus of b-catenin (present in 12–26% of HCCs),

21

by deletions,

mutations, or epigenetic alterations of the E-cadherin gene,

and by loss-of-function mutations in the AXIN1 or AXIN2

genes (present in 8–13% of HCCs).

21,122

The capacity for

nuclear b-catenin to promote the epithelial to mesenchymal

transition has been shown to induce invasive and metastatic

(5)

properties of tumor cells.

123

Correspondingly, the silencing of

b-catenin reverses the epithelial–mesenchymal transition and

represses metastatic potential.

124

In a recent review of

miRNA regulation of the Wnt/b-catenin pathway,

125

it is

apparent that miR-21, miR-200, miR-315, and miR-135

directly regulate the Wnt/b-catenin core. MiR-135 and

miR-315 activate b-catenin by inhibiting the negative regulators,

APC and axin, respectively.

126,127

MiR-200a has been shown

to regulate b-catenin levels either directly

128

or indirectly by

modulating ZEB1/2, and this downregulation of miR-200a in

HCC has been reported in various studies,

4,65,106,129,130

thereby supporting its role in mediating an increase in nuclear

b-catenin levels and the activation of the pathway in cancer

cells. MiR-34a has also been shown to be a negative regulator

of the Wnt pathway based on its targeting of WNT1.

131,132

In

HCC where miR-34 is downregulated, repression of WNT1 is

released. Moreover, in addition to invasion and metastasis,

the Wnt/b-catenin pathway is also important for maintaining

stemness.

118–120

In this context, it has been proposed that

upregulation of miR-181b supports this function at two levels,

by repressing the Wnt/b-catenin inhibitor, NLK, and by

blocking the hepatic differentiation transcription factors,

CDX2 and GATA6,

133

suggesting that liver cancer cells may

arise from stem cells that have lost control over their

self-renewal potential.

As described above, miRNAs appear to have an essential

role in the modulation of complex cross-talk that exists

between

pathways

affecting

HCC

development

and

progression. Their understanding may facilitate the

develop-ment of novel, targeted therapeutic strategies against HCC.

Animal Models of Liver Cancer

The importance of deregulated miRNAs in malignant cell

transformation and tumor development has been observed in

several studies. In particular, the development of animal

models that are genetically modified at miRNA loci have

proven the involvement of miRNAs in the initiation and

progression of cancers, and provided in vivo models to test

the efficacy of miRNA-based therapeutic approaches. Most of

the models that have been developed are related to

hematopoietic diseases, but HCC models have also been

developed (Table 1).

miRNA-specific models of HCC. For HCC, various cancer

genes, including tumor suppressor genes, oncogenes, and

hepatitis B virus or hepatitis C virus (HCV) viral genes, have

been employed for the development of mouse models.

134–144

More recently, miRNA-based mouse models predisposed to

HCC have been reported. A transgenic mouse model

characterized by overexpression of miR-221 in the liver

was developed. In these mice, male animals exhibit a strong

predisposition for HCC, which includes the emergence of

spontaneous nodular liver lesions with age and a strong

acceleration of tumor development after treatment with the

carcinogen,

N-nitrosodiethylamine

(DEN).

Notably,

the

Figure 3

Aberrant miRNA expression in liver cancer supports the activation of various signaling pathways. Simplified diagrams of RAS/MAPK, PI3K/AKT/mTOR, and WNT/b-catenin signaling pathways are shown, along with the effects of various miRNAs that are aberrantly expressed in liver cancer. As a result, cell proliferation, survival, and maintenance of stemness are enhanced

(6)

TG221 mouse was the first transgenic animal model to

develop a solid tumor following overexpression of a

miRNA.

145

Among the miRNAs that are downregulated in

HCC, two studies have reported the development of miR-122

knockout mice. These mice are characterized by hepatic

inflammation, fibrosis, and the development of spontaneous

liver tumors with age. These studies also demonstrated the

tumor suppressor function of miR-122 in the liver, as well as

its

importance

in

liver

metabolism

and

hepatocyte

differentiation.

146,147

In addition to these miRNA-specific models, the importance

of deregulated miRNA machinery components has also

recently been suggested. For DDX20, a DEAD box protein

component of miRNA-containing ribonucleoprotein

com-plexes, it suppresses NF-kB activity by regulating miR-140

function.

148

Correspondingly, miR-140 knockout mice

149

are

prone to HCC after treatment with DEN compared with control

animals, and they have a phenotype that is similar to that for

DDX20 deficiency. Taken together, these results suggest that

miR-140 may act as a tumor suppressor by regulating the

NF-kB pathway.

149

These reports confirm the importance of

miRNA deregulation in liver cancer.

Traditional HCC preclinical models. In addition to

geneti-cally modified mouse models, traditional preclinical models of

liver cancer have been established with chemical treatment.

There are two main methods of HCC induction using

chemicals: (i) administration of carcinogens alone, such as

DEN, over a prolonged period of time or by intraperitoneum

injection,

150

or (ii) administration of carcinogens (CCl

4

and

DEN) followed by a promotion phase (partial hepatectomy) or

administration of alcohol or phenobarbital.

151,152

DEN is

frequently used as a carcinogenic agent;

152

however, the

target organ in which it induces malignant tumors is species

specific. Thus, mice treated with DEN develop liver tumors as

well as gastrointestinal,

153

skin, lung,

154

and haematopoietic

malignancies.

155

In addition, the time needed to develop HCC

after a single DEN injection depends on the administered

dose, and the gender, age, and strain of the treated mice.

156

Other models of chemically induced liver disease involve

the administration of CCl

4

in drinking water, by inhalation, or

by subcutaneous or intraperitoneal injections.

151,157

CCl

4

treatment has been shown to induce an advanced stage of

liver cirrhosis and related complications, such as ascites

decompensation, in both rats and mice.

157,158

However,

development of HCC has not been reported. In a two-stage

chemical model of HCC, CCl

4

administration is accompanied

by alcohol supplementation in drinking water.

159

However,

only after 104 weeks does this combination induce the

development of HCC and metastases.

miRNA-Based Therapies

A comparative analysis of global gene expression patterns for

murine and human HCC has shown that many models can

reproduce key biological and molecular events observed in

the human condition.

160

Accordingly, mouse models have

been widely used for the testing of potential therapeutic

targets and for preclinical studies (see Li et al.

161

for a recent

review). The efficacy of miRNA-mediated HCC prevention

and therapy has also been evaluated in recent years using

various strategies involving miRNA inhibition or miRNA

replacement.

miRNA inhibition. Krutzfeldt and colleagues were the first

to demonstrate that miRNAs could be effectively silenced in

mice using specific molecules. In this study, the liver-specific

expression of miR-122 was inhibited with an intravenous

administration of chemically modified single-stranded RNA,

known as ‘antagomirs’, that were complementary to the

target miRNA.

162

The safety of this silencing approach was

established in nonhuman primates, where no evidence of

liver toxicity was detected.

163

On the basis of these

promising results, many studies of miRNA inhibition were

subsequently performed. The tumor-promoting activity of

miR-221 in HCC was also demonstrated in vivo. In one

notable study, systemic delivery of a cholesterol-tagged

anti-miR-221 to orthotopic HCC tumors was found to reduce

tumor cell proliferation and promote survival.

164

In a second

study, anti-miR-221 oligonucleotides were found to exhibit

significant antitumor activity in the TG221 transgenic mouse

model, where a significant reduction in the number and size

of tumors in the treated mice was confirmed with

histopatho-logical analyses. Significant downregulation of miR-221

levels has also been detected in liver tissues of

anti-miR-221-treated mice, thereby confirming the ability of these

molecules to inhibit endogenous miR-221

145

(Table 2). The

importance of manipulating miRNA levels as a therapeutic

tool for treating liver cancer is also supported in the work by

Lim et al.,

143

where anti-miR inhibition of miR-494 (a miRNA

Table 1 miRNA-based animal models

miRNA Mouse model name Mouse model Tumor

miR-155 E(mu)-mmu-miR155 Transgenic B-cell malignancies miR-155 mir-155LSLtTA Knockin B-cell malignancies miR-29a/b Emu-miR-29 Transgenic B-cell malignancies

miR15a/16-1 DLEU2/miR15a/16-1 Knockout Chronic lymphocytic leukemia miR-21 mir-21LSL-Tetoff Knockout B-cell malignancies

miR-125b Emu/miR-125b Transgenic B-cell malignancies miR-146a miR-146a KO Knockout Myeloproliferative disease miR-17B92 Emu-miR-17B92 Transgenic B-cell malignancies miR-122 Mir122-LKO Knockout Hepatocellular carcinoma miR-122 Mir122a–/– Knockout Hepatocellular carcinoma miR-140 MiRNA-140 /  Knockout Hepatocellular carcinoma

(7)

overexpressed in human HCC) resulted in a significant

reduction in tumor size in a primary MYC-driven liver tumor

mouse model.

HCV promotes HCC by inducing chronic liver disease and

cirrhosis. MiR-122 has been found to support HCV

replica-tion.

165–167

Correspondingly,

treatment

of

chronically

infected nonhuman primates with an LNA molecule

comple-mentary to the miR-122 sequence led to a long-lasting and

well-tolerated suppression of HCV viremia. These results

support the hypothesis that miR-122 represents a potential

target for antiviral intervention and possible liver disease

sequels.

168,169

Currently, an anti-miR-122 oligonucleotide,

‘miravirsen SPC3649’ (Santaris Pharma, A/S, Hørsholm,

Denmark), is being evaluated in a phase 2 clinical trial for the

treatment of HCV infection (ClinicalTrials.gov Identifier

NCT01200420)

170

(Figure 4). However, the concomitant

inhibition of the tumor-suppressor activity of miR-122

65,146,147

must also be considered.

miRNA replacement. The miRNA restoration is another

strategy that has been used to elucidate the functions of

miRNAs, to identify their roles in tumorigenesis, and to

provide preclinical tools for the treatment of cancer. One

anticancer approach involves the restoration of miRNAs that

are generally downregulated in cancer cells. In vivo xenograft

models have been used to test the tumor suppressor activity

of several miRNAs that were delivered by adeno-associated

viruses (AAV), nano-sized lipid particles, or by

cholesterol-conjugated modified oligonucleotides (Table 2). Hou et al.

171

used both cholesterol-conjugated small RNAs and an AAV

delivery system to effectively restore miR-199a/b-3p

expres-sion in HCC tissues of HCC-bearing nude mice. As a result,

tumor growth was inhibited without evidence of toxicity.

Lentivirus-mediated expression of miR-199a was also able to

inhibit tumor growth in a nude mouse xenograft model,

172

thereby confirming the antitumor effect of this miRNA in HCC

and its therapeutic potential.

To provide a better model of human HCC, genetically

engineered or chemically induced mouse models have been

employed. For example, using MYC-driven mouse models of

HCC, the tumor suppressor activity of miR-26a

26

and

miR-122

146

have been demonstrated. In a DEN-induced HCC

mouse model, the systemic administration of miR-124 led to a

reduction in tumor growth and tumor size via induction of

apoptosis and deregulation of a miRNA/inflammatory

feed-back loop circuit involving hepatocyte nuclear factor 4alpha

(HNF4a).

173

Taken together, these studies indicate that

miRNA mimics represent a promising approach for the

treatment of cancer, and could be translated into clinical

practice. In fact, MRX34 (Mirna Therapeutics, Inc., Austin, TX,

USA), a liposome-formulated mimic of the tumor suppressor

miR-34a, is the first miRNA mimic that has entered a

multicenter phase I clinical trial aimed at evaluating safety in

patients with primary liver cancer or with liver metastasis from

other cancers (ClinicalTrials.gov Identifier: NCT01829971)

174

(Figure 4).

Oncolytic viruses. A distinct approach that has become

interlaced with miRNA expression in HCC is the field of

oncolytic viruses. These vectors have tumor-specific activity

that results in the cytolysis of cancer cells, whereas normal

tissues exhibit minimal toxicity in vivo. Thus, the use of

oncolytic viruses represents a promising approach for the

treatment of cancer (for a recent review, see Patel et al.

175

and Callegari et al.

176

). The differential expression of

miRNAs in neoplastic versus normal tissues has also been

used to improve the safety of oncolytic virus-based therapies.

For example, to overcome liver toxicity associated with the

systemic delivery of oncolytic adenoviruses, regulatory

miRNA sequences have been inserted in the genome of

conditionally replicating adenoviruses (CRAds). In one case,

CRAds regulated by miR-122 were generated to provide

liver-specific suppression of viral replication without

compro-mising the replication capacity of the modified virus in cancer

Figure 4

miRNA-based approaches that are moving toward clinical trials. An overview of the many preclinical studies that have produced promising results, as well as the clinical trials that are being planned or are underway

(8)

tissues in vivo.

177–179

Other uses of oncolytic viruses have

included the generation of a let-7-dependent adenovirus that

is able to replicate in tumor cells, and not in normal liver cells,

in a HCC xenograft model.

180

Similarly, a

miR-199-depen-dent CRAd was generated to replicate in HCC tumor cells

and not in normal liver cells. This virus was able to control

tumor growth in a subcutaneous xenograft model in nude

mice, and in HCCs arising in immune-competent mice,

without causing significant hepatotoxicity

181

(Table 2).

Conclusions

HCC is the most common type of liver cancer diagnosed, and

treatment options currently depend on tumor size and staging.

For patients with advanced, non-resectable HCC, the only

systemic therapy available involves the multi-kinase inhibitor,

sorafenib.

182,183

This molecule targets the tyrosine kinase

receptors VEGFR and PDGFR, as well as the serine–

threonine kinases c-RAF and BRAF. Randomized phase 3

trials with sorafenib have shown an improvement in median

overall survival and progression-free survival for HCC

patients.

182,184

However, more effective treatments that have

less toxic side effects are still needed. The use of miRNAs or

anti-miRNAs

as

antitumor

therapeutic

molecules

has

attracted a lot of attention. At present, their short-term safety

has been proven in several instances, albeit their efficacy as

anticancer agents remains to be fully confirmed. Various

studies have demonstrated that miR-26a, miR-199, miR-122,

anti-221, and 494 reduce tumor growth, and

miR-34a has entered a clinical trial. For anti-miR-122, its capacity

to inhibit HCV replication has the potential to prevent the

development of HCC. Future investigations may also focus on

the use of miRNA/anti-miRNA oligonucleotides for the

prevention of HCC owing to the limited presence, or absence,

of side effects associated with these approaches. To this

end, animal models that are predisposed to HCC represent

important instruments for testing the effectiveness of miRNA/

anti-miRNA molecules.

Animal models of HCC have been established and are

widely

used

for

preclinical

and

translational

studies.

Moreover, the study of several HCC murine models has

provided a comprehensive characterization of the molecular

mechanisms of liver carcinogenesis and has also resulted in

the development of new therapeutic strategies.

185

Compared

with human HCC, genetically modified mouse models

gen-erally develop HCC without the development of liver cirrhosis.

However, this condition is present in 480% of human HCC

cases. The administration of CCl

4

has been shown to induce

advanced stages of liver cirrhosis, although this is not

accompanied by the development of HCC.

157

Therefore,

CCl

4

treatment of genetically modified mice may lead to the

development of models that more closely represent the

human condition where HCC develops in cirrhotic livers.

Future studies will be needed to investigate these possibilities.

Acknowledgements. This work was supported by funding from the Associazione Italiana per la Ricerca sul Cancro (AIRC), the Italian Ministry of University and Research, the University of Ferrara, and by the Fondazione Cassa di Risparmio di Bologna (Carisbo).

1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116: 281–297.

2. Negrini M, Ferracin M, Sabbioni S, Croce CM. MicroRNAs in human cancer: from research to therapy. J Cell Sci 2007; 120(Pt 11): 1833–1840.

3. Gailhouste L, Ochiya T. Cancer-related microRNAs and their role as tumor suppressors and oncogenes in hepatocellular carcinoma. Histol Histopathol 2013; 28: 437–451. 4. Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T et al.

Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 2006; 25: 2537–2545.

5. Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S et al. MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology 2008; 47: 1955–1963. 6. Jemal A, Center MM, DeSantis C, Ward EM. Global patterns of cancer incidence and

mortality rates and trends. Cancer Epidemiol Biomarkers Prev 2010; 19: 1893–1907. 7. Gramantieri L, Fornari F, Callegari E, Sabbioni S, Lanza G, Croce CM et al. MicroRNA

involvement in hepatocellular carcinoma. J Cell Mol Med 2008; 12: 2189–2204. 8. Aravalli RN, Cressman EN, Steer CJ. Cellular and molecular mechanisms of

hepatocellular carcinoma: an update. Arch Toxicol 2013; 87: 227–247.

9. Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet 2012; 44: 694–698.

10. Cleary SP, Jeck WR, Zhao X, Chen K, Selitsky SR, Savich GL et al. Identification of driver genes in hepatocellular carcinoma by exome sequencing. Hepatology 2013; 58: 1693–1702.

Table 2 miRNA-based therapies in HCC preclinical models

miRNAs Mouse model Method Delivery system

miR-221 Orthotopic model MiRNA inhibition Cholesterol-conjugated anti-miR-221 miR-221 TG-221 MiRNA inhibition Anti-miRNA oligonucleotide miR-494 tet-o-MYC;LAP-tTA MiRNA inhibition Anti-miRNA oligonucleotide miR-101 Xenograft MiRNA replacement RNA-duplex

miR-29b Xenograft MiRNA replacement 20-O-methyl-modified oligoribonucleotides

miR-199a/b-3p Xenograft MiRNA replacement Adeno-associated virus miR-199a/b-3p Xenograft MiRNA replacement Lentivirus

miR-375 Xenograft MiRNA replacement Cholesterol-conjugated 20-O-methyl-modified miRNA

miR-26a tet-o-MYC;LAP-tTA MiRNA replacement Adeno-associated virus miR-122 tet-o-MYC;LAP-tTAT; MiRNA replacement Adeno-associated virus miR-122 Xenograft MiRNA replacement Cationic lipid nanoparticles miR-124 DEN-induced HCC mouse model MiRNA replacement Liposomes

Let-7 Xenograft Oncolytic virus Adenovirus miR-122 Xenograft Oncolytic virus Adenovirus miR-122;

miR-124; let-7

Xenograft Oncolytic virus Herpes simplex virus miR-199 Xenograft; TG-221 Oncolytic virus Adenovirus

(9)

11. Kan Z, Zheng H, Liu X, Li S, Barber TD, Gong Z et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res 2013; 23: 1422–1433.

12. Villanueva A, Llovet JM. Targeted therapies for hepatocellular carcinoma. Gastroenterology 2011; 140: 1410–1426.

13. Faivre S, Bouattour M, Raymond E. Novel molecular therapies in hepatocellular carcinoma. Liver Int 2011; 31(Suppl 1): 151–160.

14. Negrini M, Gramantieri L, Sabbioni S, Croce CM. microRNA involvement in hepatocellular carcinoma. Anticancer Agents Med Chem 2011; 11: 500–521.

15. Scarola M, Schoeftner S, Schneider C, Benetti R. miR-335 directly targets Rb1 (pRb/p105) in a proximal connection to p53-dependent stress response. Cancer Res 2010; 70: 6925–6933.

16. Tome M, Lopez-Romero P, Albo C, Sepulveda JC, Fernandez-Gutierrez B, Dopazo A et al. miR-335 orchestrates cell proliferation, migration and differentiation in human mesenchymal stem cells. Cell Death Differ 2010; 18: 985–995.

17. Lupini L, Bassi C, Ferracin M, Bartonicek N, D’Abundo L, Zagatti B et al. miR-221 affects multiple cancer pathways by modulating the level of hundreds messenger RNAs. Front Genet 2013; 4: 64.

18. Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, Calin GA et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene 2008; 27: 5651–5661.

19. Xu XR, Huang J, Xu ZG, Qian BZ, Zhu ZD, Yan Q et al. Insight into hepatocellular carcinogenesis at transcriptome level by comparing gene expression profiles of hepatocellular carcinoma with those of corresponding noncancerous liver. Proc Natl Acad Sci USA 2001; 98: 15089–15094.

20. Joo M, Kang YK, Kim MR, Lee HK, Jang JJ. Cyclin D1 overexpression in hepatocellular carcinoma. Liver 2001; 21: 89–95.

21. Laurent-Puig P, Legoix P, Bluteau O, Belghiti J, Franco D, Binot F et al. Genetic alterations associated with hepatocellular carcinomas define distinct pathways of hepatocarcinogenesis. Gastroenterology 2001; 120: 1763–1773.

22. Matsuda Y, Ichida T, Matsuzawa J, Sugimura K, Asakura H. p16(INK4) is inactivated by extensive CpG methylation in human hepatocellular carcinoma. Gastroenterology 1999; 116: 394–400.

23. Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I, Inazawa J. miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 2010; 31: 766–776.

24. Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP, Zhuang SM. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology 2009; 50: 113–121.

25. Ji J, Shi J, Budhu A, Yu Z, Forgues M, Roessler S et al. MicroRNA expression, survival, and response to interferon in liver cancer. N Engl J Med 2009; 361: 1437–1447.

26. Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang HW et al. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell 2009; 137: 1005–1017.

27. Ito Y, Takeda T, Sakon M, Tsujimoto M, Monden M, Matsuura N. Expression of p57/Kip2 protein in hepatocellular carcinoma. Oncology 2001; 61: 221–225.

28. Schwienbacher C, Gramantieri L, Scelfo R, Veronese A, Calin GA, Bolondi L et al. Gain of imprinting at chromosome 11p15: A pathogenetic mechanism identified in human hepatocarcinomas. Proc Natl Acad Sci USA 2000; 97: 5445–5449.

29. Li Y, Tan W, Neo TW, Aung MO, Wasser S, Lim SG et al. Role of the miR-106b-25 microRNA cluster in hepatocellular carcinoma. Cancer Sci 2009; 100: 1234–1242.

30. Petrocca F, Visone R, Onelli MR, Shah MH, Nicoloso MS, de Martino I et al. E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 2008; 13: 272–286.

31. Carraro G, El-Hashash A, Guidolin D, Tiozzo C, Turcatel G, Young BM et al. miR-17 family of microRNAs controls FGF10-mediated embryonic lung epithelial branching morphogenesis through MAPK14 and STAT3 regulation of E-Cadherin distribution. Dev Biol 2009; 333: 238–250.

32. O’Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005; 435: 839–843.

33. Taguchi A, Yanagisawa K, Tanaka M, Cao K, Matsuyama Y, Goto H et al. Identification of hypoxia-inducible factor-1 alpha as a novel target for miR-17-92 microRNA cluster. Cancer Res 2008; 68: 5540–5545.

34. Connolly E, Melegari M, Landgraf P, Tchaikovskaya T, Tennant BC, Slagle BL et al. Elevated expression of the miR-17-92 polycistron and miR-21 in hepadnavirus-associated hepatocellular carcinoma contributes to the malignant phenotype. Am J Pathol 2008; 173: 856–864.

35. Fabregat I, Roncero C, Fernandez M. Survival and apoptosis: a dysregulated balance in liver cancer. Liver Int 2007; 27: 155–162.

36. Guo XZ, Shao XD, Liu MP, Xu JH, Ren LN, Zhao JJ et al. Effect of bax, bcl-2 and bcl-xL on regulating apoptosis in tissues of normal liver and hepatocellular carcinoma. World J Gastroenterol 2002; 8: 1059–1062.

37. Lin CJ, Gong HY, Tseng HC, Wang WL, Wu JL. miR-122 targets an anti-apoptotic gene, Bcl-w, in human hepatocellular carcinoma cell lines. Biochem Biophys Res Commun 2008; 375: 315–320.

38. Shimizu S, Takehara T, Hikita H, Kodama T, Miyagi T, Hosui A et al. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. J Hepatol 2010; 52: 698–704.

39. Su H, Yang JR, Xu T, Huang J, Xu L, Yuan Y et al. MicroRNA-101, down-regulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res 2009; 69: 1135–1142.

40. Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology 2010; 51: 881–890.

41. Mott JL, Kobayashi S, Bronk SF, Gores GJ. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene 2007; 26: 6133–6140.

42. Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH et al. Effects of microRNA-29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology 2010; 51: 836–845.

43. Gramantieri L, Fornari F, Ferracin M, Veronese A, Sabbioni S, Calin GA et al. MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality. Clin Cancer Res 2009; 15: 5073–5081.

44. Schmelzle T, Mailleux AA, Overholtzer M, Carroll JS, Solimini NL, Lightcap ES et al. Functional role and oncogene-regulated expression of the BH3-only factor Bmf in mammary epithelial anoikis and morphogenesis. Proc Natl Acad Sci USA 2007; 104: 3787–3792.

45. Padua D, Massague J. Roles of TGFbeta in metastasis. Cell Res 2009; 19: 89–102. 46. Fu H, Tie Y, Xu C, Zhang Z, Zhu J, Shi Y et al. Identification of human fetal liver miRNAs

by a novel method. FEBS Lett 2005; 579: 3849–3854.

47. Veronese A, Lupini L, Consiglio J, Visone R, Ferracin M, Fornari F et al. Oncogenic role of miR-483-3p at the IGF2/483 locus. Cancer Res 2010; 70: 3140–3149.

48. Saini S, Majid S, Yamamura S, Tabatabai ZL, Suh SO, Shahryari V et al. Regulatory role of miR-203 in prostate cancer progression and metastasis. Clin Cancer Res 2010. 49. Schetter AJ, Leung SY, Sohn JJ, Zanetti KA, Bowman ED, Yanaihara N et al. MicroRNA

expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA 2008; 299: 425–436.

50. Li R, You N, Wang X, Dou K. MicroRNAs involved in neoplastic transformation of liver cancer stem cells. J Exp Clin Cancer Res 2010; 29: 169.

51. Meng F, Henson R, Lang M, Wehbe H, Maheshwari S, Mendell JT et al. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarci-noma cell lines. Gastroenterology 2006; 130: 2113–2129.

52. Blower PE, Chung JH, Verducci JS, Lin S, Park JK, Dai Z et al. MicroRNAs modulate the chemosensitivity of tumor cells. Mol Cancer Ther 2008; 7: 1–9.

53. Tomimaru Y, Eguchi H, Nagano H, Wada H, Tomokuni A, Kobayashi S et al. MicroRNA-21 induces resistance to the anti-tumour effect of interferon-alpha/ 5-fluorouracil in hepatocellular carcinoma cells. Br J Cancer 2010; 103: 1617–1626.

54. Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007; 133: 647–658.

55. Sayed D, He M, Hong C, Gao S, Rane S, Yang Z et al. MicroRNA-21 is a downstream effector of AKT that mediates its antiapoptotic effects via suppression of Fas ligand. J Biol Chem 2010; 285: 20281–20290.

56. Zhang JG, Wang JJ, Zhao F, Liu Q, Jiang K, Yang GH. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin Chim Acta 2010; 411: 846–852.

57. Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008; 27: 2128–2136.

58. Chen Y, Liu W, Chao T, Zhang Y, Yan X, Gong Y et al. MicroRNA-21 down-regulates the expression of tumor suppressor PDCD4 in human glioblastoma cell T98G. Cancer Lett 2008; 272: 197–205.

59. Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem 2008; 283: 1026–1033.

60. Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH et al. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene 2008; 27: 4373–4379.

61. Motoyama K, Inoue H, Mimori K, Tanaka F, Kojima K, Uetake H et al. Clinicopathological and prognostic significance of PDCD4 and microRNA-21 in human gastric cancer. Int J Oncol 2010; 36: 1089–1095.

62. Selaru FM, Olaru AV, Kan T, David S, Cheng Y, Mori Y et al. MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3. Hepatology 2009; 49: 1595–1601. 63. Bressac B, Kew M, Wands J, Ozturk M. Selective G to T mutations of p53 gene in

hepatocellular carcinoma from southern Africa. Nature 1991; 350: 429–431. 64. Lu W, Lo SY, Chen M, Wu K, Fung YK, Ou JH. Activation of p53 tumor suppressor by

hepatitis C virus core protein. Virology 1999; 264: 134–141.

65. Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG et al. Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res 2007; 67: 6092–6099.

(10)

66. Fornari F, Gramantieri L, Giovannini C, Veronese A, Ferracin M, Sabbioni S et al. MiR-122/cyclin G1 interaction modulates p53 activity and affects doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res 2009; 69: 5761–5767.

67. Spizzo R, Nicoloso MS, Lupini L, Lu Y, Fogarty J, Rossi S et al. miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-alpha in human breast cancer cells. Cell Death Differ 2010; 17: 246–254.

68. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B et al. miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci USA 2010; 107: 264–269.

69. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007; 26: 745–752.

70. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y et al. A microRNA component of the p53 tumour suppressor network. Nature 2007; 447: 1130–1134.

71. He X, He L, Hannon GJ. The guardian’s little helper: microRNAs in the p53 tumor suppressor network. Cancer Res 2007; 67: 11099–11101.

72. Hermeking H. p53 enters the microRNA world. Cancer Cell 2007; 12: 414–418. 73. Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N et al.

Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell 2007; 26: 731–743.

74. Tarasov V, Jung P, Verdoodt B, Lodygin D, Epanchintsev A, Menssen A et al. Differential regulation of microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest. Cell Cycle 2007; 6: 1586–1593.

75. Sachdeva M, Zhu S, Wu F, Wu H, Walia V, Kumar S et al. p53 represses c-Myc through induction of the tumor suppressor miR-145. Proc Natl Acad Sci USA 2009; 106: 3207–3212.

76. Georges SA, Biery MC, Kim SY, Schelter JM, Guo J, Chang AN et al. Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res 2008; 68: 10105–10112.

77. Braun CJ, Zhang X, Savelyeva I, Wolff S, Moll UM, Schepeler T et al. p53-Responsive micrornas 192 and 215 are capable of inducing cell cycle arrest. Cancer Res 2008; 68: 10094–10104.

78. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol 2003; 4: 915–925.

79. Benvenuti S, Comoglio PM. The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol 2007; 213: 316–325.

80. Boix L, Rosa JL, Ventura F, Castells A, Bruix J, Rodes J et al. c-met mRNA overexpression in human hepatocellular carcinoma. Hepatology 1994; 19: 88–91. 81. Suzuki K, Hayashi N, Yamada Y, Yoshihara H, Miyamoto Y, Ito Y et al. Expression

of the c-met protooncogene in human hepatocellular carcinoma. Hepatology 1994; 20: 1231–1236.

82. Grigioni WF, Fiorentino M, D’Errico A, Ponzetto A, Crepaldi T, Prat M et al. Overexpression of c-met protooncogene product and raised Ki67 index in hepatocellular carcinomas with respect to benign liver conditions. Hepatology 1995; 21: 1543–1546.

83. Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor, the c-met proto-oncogene, in hepatocellular carcinoma. Hepatology 1997; 25: 619–623.

84. Fornari F, Milazzo M, Chieco P, Negrini M, Calin GA, Grazi GL et al. MiR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res 2010; 70: 5184–5193.

85. Kim S, Lee UJ, Kim MN, Lee EJ, Kim JY, Lee MY et al. MicroRNA miR-199a* regulates the MET proto-oncogene and the downstream extracellular signal-regulated kinase 2 (ERK2). J Biol Chem 2008; 283: 18158–18166.

86. Li N, Fu H, Tie Y, Hu Z, Kong W, Wu Y et al. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett 2009; 275: 44–53.

87. Salvi A, Sabelli C, Moncini S, Venturin M, Arici B, Riva P et al. MicroRNA-23b mediates urokinase and c-met downmodulation and a decreased migration of human hepatocellular carcinoma cells. FEBS J 2009; 276: 2966–2982.

88. Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, Majumder S et al. Methylation mediated silencing of MicroRNA-1 gene and its role in hepatocellular carcinogenesis. Cancer Res 2008; 68: 5049–5058.

89. Jagirdar J, Nonomura A, Patil J, Thor A, Paronetto F. ras oncogene p21 expression in hepatocellular carcinoma. J Exp Pathol 1989; 4: 37–46.

90. Challen C, Guo K, Collier JD, Cavanagh D, Bassendine MF. Infrequent point mutations in codons 12 and 61 of ras oncogenes in human hepatocellular carcinomas. J Hepatol 1992; 14: 342–346.

91. Akao Y, Nakagawa Y, Naoe T. let-7 microRNA functions as a potential growth suppressor in human colon cancer cells. Biol Pharm Bull 2006; 29: 903–906.

92. Bussing I, Slack FJ, Grosshans H. let-7 microRNAs in development, stem cells and cancer. Trends Mol Med 2008; 14: 400–409.

93. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A et al. RAS is regulated by the let-7 microRNA family. Cell 2005; 120: 635–647.

94. Lee YS, Dutta A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev 2007; 21: 1025–1030.

95. Mayr C, Hemann MT, Bartel DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 2007; 315: 1576–1579.

96. Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res 2004; 64: 3753–3756.

97. Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 2007; 131: 1109–1123.

98. Liu C, Yu J, Yu S, Lavker RM, Cai L, Liu W et al. MicroRNA-21 acts as an oncomir through multiple targets in human hepatocellular carcinoma. J Hepatol 2010; 53: 98–107. 99. Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A et al. miR-221&222

regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 2009; 16: 498–509.

100. Connolly EC, Van Doorslaer K, Rogler LE, Rogler CE. Overexpression of miR-21 promotes an in vitro metastatic phenotype by targeting the tumor suppressor RHOB. Mol Cancer Res 2010; 8: 691–700.

101. Gabriely G, Wurdinger T, Kesari S, Esau CC, Burchard J, Linsley PS et al. MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol 2008; 28: 5369–5380.

102. Song G, Sharma AD, Roll GR, Ng R, Lee AY, Blelloch RH et al. MicroRNAs control hepatocyte proliferation during liver regeneration. Hepatology 2010; 51: 1735–1743. 103. Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo YY. MicroRNA-21 targets tumor suppressor

genes in invasion and metastasis. Cell Res 2008; 18: 350–359.

104. Villanueva A, Chiang DY, Newell P, Peix J, Thung S, Alsinet C et al. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology 2008; 135: 1972–1983; 1983 e1-11.

105. Zhou L, Huang Y, Li J, Wang Z. The mTOR pathway is associated with the poor prognosis of human hepatocellular carcinoma. Med Oncol 2010; 27: 255–261.

106. Jiang J, Gusev Y, Aderca I, Mettler TA, Nagorney DM, Brackett DJ et al. Association of MicroRNA expression in hepatocellular carcinomas with hepatitis infection, cirrhosis, and patient survival. Clin Cancer Res 2008; 14: 419–427.

107. Shen Q, Cicinnati VR, Zhang X, Iacob S, Weber F, Sotiropoulos GC et al. Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol Cancer 2010; 9: 227.

108. Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K et al. Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int J Cancer 2009; 125: 345–352.

109. Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casalini P et al. MicroRNA signatures in human ovarian cancer. Cancer Res 2007; 67: 8699–8707.

110. Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007; 129: 1401–1414.

111. Dolganiuc A, Petrasek J, Kodys K, Catalano D, Mandrekar P, Velayudham A et al. MicroRNA expression profile in Lieber-DeCarli diet-induced alcoholic and methionine choline deficient diet-induced nonalcoholic steatohepatitis models in mice. Alcohol Clin Exp Res 2009; 33: 1704–1710.

112. Chao A, Tsai CL, Wei PC, Hsueh S, Chao AS, Wang CJ et al. Decreased expression of microRNA-199b increases protein levels of SET (protein phosphatase 2A inhibitor) in human choriocarcinoma. Cancer Lett 2010; 291: 99–107.

113. Coulouarn C, Factor VM, Andersen JB, Durkin ME, Thorgeirsson SS. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene 2009; 28: 3526–3536.

114. Tsai WC, Hsu PW, Lai TC, Chau GY, Lin CW, Chen CM et al. MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology 2009; 49: 1571–1582.

115. Wong CC, Wong CM, Tung EK, Au SL, Lee JM, Poon RT et al. The MicroRNA miR-139 Suppresses Metastasis and Progression of Hepatocellular Carcinoma by Down-regulating Rho-Kinase 2. Gastroenterology 2011; 140: 322–331.

116. Ding J, Huang S, Wu S, Zhao Y, Liang L, Yan M et al. Gain of miR-151 on chromosome 8q24.3 facilitates tumour cell migration and spreading through downregulating RhoGDIA. Nat Cell Biol 2010; 12: 390–399.

117. Polakis P. Wnt signaling and cancer. Genes Dev 2000; 14: 1837–1851.

118. Fodde R, Brabletz T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr Opin Cell Biol 2007; 19: 150–158.

119. Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol 2010; 12: 468–476.

120. Wend P, Holland JD, Ziebold U, Birchmeier W. Wnt signaling in stem and cancer stem cells. Semin Cell Dev Biol 2010; 21: 855–863.

121. Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease. Hepatology 2007; 45: 1298–1305.

122. Satoh S, Daigo Y, Furukawa Y, Kato T, Miwa N, Nishiwaki T et al. AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1. Nat Genet 2000; 24: 245–250.

123. Brabletz T, Hlubek F, Spaderna S, Schmalhofer O, Hiendlmeyer E, Jung A et al. Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and beta-catenin. Cells Tissues Organs 2005; 179: 56–65.

Figura

Figure 1 Aberrant miRNA expression in liver cancer promotes cell-cycle progression. A simplified pathway of cell cycle regulation is presented that shows the effects of various aberrantly expressed miRNAs in liver cancer
Figure 4 miRNA-based approaches that are moving toward clinical trials. An overview of the many preclinical studies that have produced promising results, as well as the clinical trials that are being planned or are underway
Table 2 miRNA-based therapies in HCC preclinical models

Riferimenti

Documenti correlati

Validavimo kohor- toje 21-os mikro RNR raiška nustatyta skrandžio vėžio (n = 39) ir 21-os atrofinio gastrito audinyje (n = 40) palyginti su kontroliniu skrandžio

[34] provided a list of guidelines for Design for Reliability (DfR) which are (1) simplicity, (2) use of proven components and preferred designs, (3) stress and strength design, (4)

In order to assess the importance of the heating induced by the synchrotron radiation in the present experiment, we developed a 3D finite element model (FEM)

In these settings, both overcrowding and high detainee turnover of individuals with blood-borne infections and tuberculosis often restricts screening, prevention, and

Generally, compared to non-Aboriginal inmates, a greater proportion of Aboriginal inmates indicated they were not willing to be tested because of fear of the test result, and

The research of this paper was supported by INDAM-GNSAGA, and by the project NRF (South Africa) with the grant ITAL170904261537, Ref. The author thanks the anonymous referees for

Many literature data showed a benefit from prophylactic antiviral treatment in cancer patients at risk for HBV reactivation, however there is no evidence in determining the benefit

High Sustained Virologic Response Rates in Liver Transplant Recipients With Recurrent HCV Genotype 1 Infection Receiving ABT-450/r/Ombitasvir Dasabuvir Plus Ribavirin. 65th